Critical Role of K1685 and K1829 in the Large Protein of Rabies Virus in Viral Pathogenicity and Immune Evasion

ABSTRACT Rabies, one of the oldest infectious diseases, still presents a public health threat in most parts of the world today. Its pathogen, rabies virus (RABV), can utilize its viral proteins, such as the nucleoprotein and phosphorylation protein, to subvert the host innate immune system. For a long time, the large (L) protein was believed to be essential for RABV transcription and replication, but its role in viral pathogenicity and immune evasion was not known. Recent studies have found that the conserved K-D-K-E tetrad motif in the L protein is related to the methyltransferase (MTase) activity in the viral mRNA process. In the present study, a series of RABV mutations in this motif was constructed with the recombinant CVS-B2c (rB2c) virus. Two of these mutants, rB2c-K1685A and rB2c-K1829A, were found to be stable and displayed an attenuated phenotype in both in vitro growth and in vivo pathogenicity in adult and suckling mice. Further studies demonstrated that these two mutants were more sensitive to the expression of the interferon-stimulated gene product IFIT2 than the parent virus. Taken together, our results suggest that K1685 and K1829 in the L protein play important roles in pathogenicity and immune evasion during RABV infection. IMPORTANCE Rabies continues to present a public health threat in most areas of the world, especially in the developing countries of Asia and Africa. The pathogenic mechanisms for rabies are not well understood. In the present study, it was found that the recombinant rabies viruses rB2c-K1685A and rB2c-K1829A, carrying mutations at the predicted MTase catalytic sites in the L protein, were highly attenuated both in vitro and in vivo. Further studies showed that these mutants were more sensitive to the expression of the interferon-stimulated gene product IFIT2 than the parent virus. These findings improve our understanding of rabies pathogenesis, which may help in developing potential therapeutics and an avirulent rabies vaccine.

[1]  Bo Liang,et al.  Structure of the L Protein of Vesicular Stomatitis Virus from Electron Cryomicroscopy , 2015, Cell.

[2]  Lisa E. Gralinski,et al.  Attenuation and Restoration of Severe Acute Respiratory Syndrome Coronavirus Mutant Lacking 2′-O-Methyltransferase Activity , 2014, Journal of Virology.

[3]  S. Whelan,et al.  mRNA Cap Methylation Influences Pathogenesis of Vesicular Stomatitis Virus In Vivo , 2013, Journal of Virology.

[4]  David C. Chang,et al.  Correction: Rational Design of a Live Attenuated Dengue Vaccine: 2′-O-Methyltransferase Mutants Are Highly Attenuated and Immunogenic in Mice and Macaques , 2013, PLoS Pathogens.

[5]  K. Takeda,et al.  Ifit1 Inhibits Japanese Encephalitis Virus Replication through Binding to 5′ Capped 2′-O Unmethylated RNA , 2013, Journal of Virology.

[6]  Hualei Wang,et al.  Rabies virus glycoprotein is an important determinant for the induction of innate immune responses and the pathogenic mechanisms. , 2013, Veterinary microbiology.

[7]  Xuping Xie,et al.  Rational Design of a Flavivirus Vaccine by Abolishing Viral RNA 2′-O Methylation , 2013, Journal of Virology.

[8]  M. Diamond,et al.  The broad-spectrum antiviral functions of IFIT and IFITM proteins , 2012, Nature Reviews Immunology.

[9]  M. Diamond,et al.  Interferon-Induced Ifit2/ISG54 Protects Mice from Lethal VSV Neuropathogenesis , 2012, PLoS pathogens.

[10]  E. Decroly,et al.  Conventional and unconventional mechanisms for capping viral mRNA , 2011, Nature Reviews Microbiology.

[11]  Ying Sun,et al.  Biochemical and Structural Insights into the Mechanisms of SARS Coronavirus RNA Ribose 2′-O-Methylation by nsp16/nsp10 Protein Complex , 2011, PLoS pathogens.

[12]  Gérard Bricogne,et al.  Crystal Structure and Functional Analysis of the SARS-Coronavirus RNA Cap 2′-O-Methyltransferase nsp10/nsp16 Complex , 2011, PLoS pathogens.

[13]  G. Sen,et al.  The ISG56/IFIT1 gene family. , 2011, Journal of interferon & cytokine research : the official journal of the International Society for Interferon and Cytokine Research.

[14]  Burkhard Ludewig,et al.  Ribose 2′-O-methylation provides a molecular signature for the distinction of self and non-self mRNA dependent on the RNA sensor Mda5 , 2011, Nature Immunology.

[15]  Hongping Dong,et al.  2′-O methylation of the viral mRNA cap evades host restriction by IFIT family members , 2010, Nature.

[16]  K. Conzelmann,et al.  Genetic Dissection of Interferon-Antagonistic Functions of Rabies Virus Phosphoprotein: Inhibition of Interferon Regulatory Factor 3 Activation Is Important for Pathogenicity , 2010, Journal of Virology.

[17]  M. Gale,et al.  Rabies Virus Infection Induces Type I Interferon Production in an IPS-1 Dependent Manner While Dendritic Cell Activation Relies on IFNAR Signaling , 2010, PLoS pathogens.

[18]  Hualei Wang,et al.  Expression of MIP-1α (CCL3) by a Recombinant Rabies Virus Enhances Its Immunogenicity by Inducing Innate Immunity and Recruiting Dendritic Cells and B Cells , 2010, Journal of Virology.

[19]  Haitao Guo,et al.  Identification of Five Interferon-Induced Cellular Proteins That Inhibit West Nile Virus and Dengue Virus Infections , 2010, Journal of Virology.

[20]  N. Ito,et al.  Rabies Virus Nucleoprotein Functions To Evade Activation of the RIG-I-Mediated Antiviral Response , 2010, Journal of Virology.

[21]  K. Conzelmann,et al.  Rhabdovirus evasion of the interferon system. , 2009, Journal of interferon & cytokine research : the official journal of the International Society for Interferon and Cytokine Research.

[22]  Huanchun Chen,et al.  The Roles of Chemokines in Rabies Virus Infection: Overexpression May Not Always Be Beneficial , 2009, Journal of Virology.

[23]  Z. Fu,et al.  Role of chemokines in the enhancement of BBB permeability and inflammatory infiltration after rabies virus infection. , 2009, Virus research.

[24]  D. Hooper,et al.  Immune evasion by rabies viruses through the maintenance of blood-brain barrier integrity , 2008, Journal of NeuroVirology.

[25]  Bryan R. G. Williams,et al.  Interferon-inducible antiviral effectors , 2008, Nature Reviews Immunology.

[26]  S. Whelan,et al.  A Conserved Motif in Region V of the Large Polymerase Proteins of Nonsegmented Negative-Sense RNA Viruses That Is Essential for mRNA Capping , 2007, Journal of Virology.

[27]  Yi Guo,et al.  Structure and Function of Flavivirus NS5 Methyltransferase , 2007, Journal of Virology.

[28]  H. Koprowski,et al.  Failure To Open the Blood-Brain Barrier and Deliver Immune Effectors to Central Nervous System Tissues Leads to the Lethal Outcome of Silver-Haired Bat Rabies Virus Infection , 2006, Journal of Virology.

[29]  I. Campbell,et al.  Coordinated Regulation and Widespread Cellular Expression of Interferon-Stimulated Genes (ISG) ISG-49, ISG-54, and ISG-56 in the Central Nervous System after Infection with Distinct Viruses , 2006, Journal of Virology.

[30]  Yi Guo,et al.  West Nile Virus 5′-Cap Structure Is Formed by Sequential Guanine N-7 and Ribose 2′-O Methylations by Nonstructural Protein 5 , 2006, Journal of Virology.

[31]  K. Honda,et al.  Type I Inteferon Gene Induction by the Interferon Regulatory Factor Family of Transcription Factors , 2006 .

[32]  S. Whelan,et al.  A unique strategy for mRNA cap methylation used by vesicular stomatitis virus. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[33]  S. Akira,et al.  Pathogen Recognition and Innate Immunity , 2006, Cell.

[34]  D. Blondel,et al.  Rabies Virus P Protein Interacts with STAT1 and Inhibits Interferon Signal Transduction Pathways , 2005, Journal of Virology.

[35]  S. Whelan,et al.  Amino Acid Residues within Conserved Domain VI of the Vesicular Stomatitis Virus Large Polymerase Protein Essential for mRNA Cap Methyltransferase Activity , 2005, Journal of Virology.

[36]  Yuhuan Wang,et al.  Attenuated Rabies Virus Activates, while Pathogenic Rabies Virus Evades, the Host Innate Immune Responses in the Central Nervous System , 2005, Journal of Virology.

[37]  S. Finke,et al.  Identification of the Rabies Virus Alpha/Beta Interferon Antagonist: Phosphoprotein P Interferes with Phosphorylation of Interferon Regulatory Factor 3 , 2005, Journal of Virology.

[38]  I. Kurane,et al.  An improved method for recovering rabies virus from cloned cDNA. , 2003, Journal of virological methods.

[39]  P. Pandolfi,et al.  Rabies virus P and small P products interact directly with PML and reorganize PML nuclear bodies , 2002, Oncogene.

[40]  B. Henrissat,et al.  Viral RNA-polymerases -- a predicted 2'-O-ribose methyltransferase domain shared by all Mononegavirales. , 2002, Trends in biochemical sciences.

[41]  Leszek Rychlewski,et al.  In silico identification, structure prediction and phylogenetic analysis of the 2'-O-ribose (cap 1) methyltransferase domain in the large structural protein of ssRNA negative-strand viruses. , 2002, Protein engineering.

[42]  A. Shatkin,et al.  Viral and cellular mRNA capping: Past and prospects , 2000, Advances in Virus Research.

[43]  N. Tordo,et al.  Sequence comparison of five polymerases (L proteins) of unsegmented negative-strand RNA viruses: theoretical assignment of functional domains. , 1990, The Journal of general virology.

[44]  C. Smith,et al.  The molecular biology of rabies viruses. , 1988, Reviews of infectious diseases.

[45]  A. J. Shatkin,et al.  5′-Terminal 7-methylguanosine in eukaryotic mRNA is required for translation , 1975, Nature.

[46]  S. Matsumoto,et al.  COMPARATIVE STUDIES BETWEEN PATHOGENESIS OF STREET AND FIXED RABIES INFECTION , 1967, Journal of Experimental Medicine.

[47]  WHO Expert Consultation on Rabies. Second report. , 2013, World Health Organization technical report series.

[48]  N. Ito,et al.  Amino acids at positions 273 and 394 in rabies virus nucleoprotein are important for both evasion of host RIG-I-mediated antiviral response and pathogenicity. , 2011, Virus research.

[49]  K. Conzelmann,et al.  Interferon in rabies virus infection. , 2011, Advances in virus research.

[50]  M. Lafon Evasive strategies in rabies virus infection. , 2011, Advances in virus research.

[51]  A. Jackson Rabies pathogenesis , 2011, Journal of NeuroVirology.

[52]  R. Ruigrok,et al.  Rabies virus transcription and replication. , 2011, Advances in virus research.

[53]  K. Honda,et al.  Type I interferon [corrected] gene induction by the interferon regulatory factor family of transcription factors. , 2006, Immunity.